Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Methods Enzymol ; 689: 201-234, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37802571

RESUMO

The 17ß-hydroxysteroid dehydrogenase type 1 (HSD17B1) has a key role in estrogen biosynthesis as it catalyzes the reduction of estrone to the most potent estrogen, estradiol. Estradiol has a high affinity for estrogen receptors and thus stimulates their transactivation, which leads to cell proliferation and numerous other effects. HSD17B2 catalyzes the oxidation of estradiol to the less potent estrone, thereby decreasing estrogen receptor activation, which results in reduction of estrogen-associated effects. HSD17B1 and HSD17B2 overexpressing E.coli homogenates or recombinant enzymes can be used for screening and development of drugs against various pathologies such as cancer, endometriosis or osteoporosis. Here we describe the preparation of HSD17B1 and HSD17B2 bacterial homogenates and purified recombinant HSD17B1 protein as enzyme sources as well as enzymatic assays based on radiometric and mass-spectrometric detection for enzyme characterization.


Assuntos
Estrogênios , Estrona , Feminino , Humanos , Estrona/metabolismo , Estrogênios/metabolismo , Estradiol/metabolismo , Ensaios Enzimáticos
2.
Metabolites ; 12(2)2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35208174

RESUMO

Naturally occurring substances are valuable resources for drug development. In this respect, chalcones are known to be antiproliferative agents against prostate cancer cell lines through various mechanisms or targets. Based on the literature and preliminary results, we aimed to study and optimise the efficiency of a series of chalcones to inhibit androgen-converting AKR1C3, known to promote prostate cancer. A total of 12 chalcones with different substitution patterns were synthesised. Structure-activity relationships associated with these modifications on AKR1C3 inhibition were analysed by performing enzymatic assays and docking simulations. In addition, the selectivity and cytotoxicity of the compounds were assessed. In enzymatic assays, C-6' hydroxylated derivatives were more active than C-6' methoxylated derivatives. In contrast, C-4 methylation increased activity over C-4 hydroxylation. Docking results supported these findings with the most active compounds fitting nicely in the binding site and exhibiting strong interactions with key amino acid residues. The most effective inhibitors were not cytotoxic for HEK293T cells and selective for 17ß-hydroxysteroid dehydrogenases not primarily involved in steroid hormone metabolism. Nevertheless, they inhibited several enzymes of the steroid metabolism pathways. Favourable substitutions that enhanced AKR1C3 inhibition of chalcones were identified. This study paves the way to further develop compounds from this series or related flavonoids with improved inhibitory activity against AKR1C3.

3.
Int J Radiat Biol ; 97(4): 529-540, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33464160

RESUMO

PURPOSE: The long-term effect of low and moderate doses of ionizing radiation on the lens is still a matter of debate and needs to be evaluated in more detail. MATERIAL AND METHODS: We conducted a detailed histological analysis of eyes from B6C3F1 mice cohorts after acute gamma irradiation (60Co source; 0.063 Gy/min) at young adult age of 10 weeks with doses of 0.063, 0.125, and 0.5 Gy. Sham irradiated (0 Gy) mice were used as controls. To test for genetic susceptibility heterozygous Ercc2 mutant mice were used and compared to wild-type mice of the same strain background. Mice of both sexes were included in all cohorts. Eyes were collected 4 h, 12, 18 and 24 months after irradiation. For a better understanding of the underlying mechanisms, metabolomics analyses were performed in lenses and plasma samples of the same mouse cohorts at 4 and 12 h as well as 12, 18 and 24 months after irradiation. For this purpose, a targeted analysis was chosen. RESULTS: This analysis revealed histological changes particularly in the posterior part of the lens that rarely can be observed by using Scheimpflug imaging, as we reported previously. We detected a significant increase of posterior subcapsular cataracts (PSCs) 18 and 24 months after irradiation with 0.5 Gy (odds ratio 9.3; 95% confidence interval 2.1-41.3) independent of sex and genotype. Doses below 0.5 Gy (i.e. 0.063 and 0.125 Gy) did not significantly increase the frequency of PSCs at any time point. In lenses, we observed a clear effect of sex and aging but not of irradiation or genotype. While metabolomics analyses of plasma from the same mice showed only a sex effect. CONCLUSIONS: This article demonstrates a significant radiation-induced increase in the incidence of PSCs, which could not be identified using Scheimpflug imaging as the only diagnostic tool.


Assuntos
Catarata/etiologia , Lesões por Radiação/etiologia , Animais , Catarata/genética , Relação Dose-Resposta à Radiação , Feminino , Heterozigoto , Cristalino/efeitos da radiação , Masculino , Camundongos , Lesões por Radiação/genética
4.
Int J Mol Sci ; 21(19)2020 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-32993084

RESUMO

Natural products comprise a rich reservoir for innovative drug leads and are a constant source of bioactive compounds. To find pharmacological targets for new or already known natural products using modern computer-aided methods is a current endeavor in drug discovery. Nature's treasures, however, could be used more effectively. Yet, reliable pipelines for the large-scale target prediction of natural products are still rare. We developed an in silico workflow consisting of four independent, stand-alone target prediction tools and evaluated its performance on dihydrochalcones (DHCs)-a well-known class of natural products. Thereby, we revealed four previously unreported protein targets for DHCs, namely 5-lipoxygenase, cyclooxygenase-1, 17ß-hydroxysteroid dehydrogenase 3, and aldo-keto reductase 1C3. Moreover, we provide a thorough strategy on how to perform computational target predictions and guidance on using the respective tools.


Assuntos
Produtos Biológicos/química , Simulação por Computador , Descoberta de Drogas , Inibidores Enzimáticos/química , Oxirredutases , Avaliação Pré-Clínica de Medicamentos , Humanos , Oxirredutases/antagonistas & inibidores , Oxirredutases/química
5.
Cancers (Basel) ; 12(8)2020 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-32731472

RESUMO

The expanded use of second-generation antiandrogens revolutionized the treatment landscape of progressed prostate cancer. However, resistances to these novel drugs are already the next obstacle to be solved. Various previous studies depicted an involvement of the enzyme AKR1C3 in the process of castration resistance as well as in the resistance to 2nd generation antiandrogens like enzalutamide. In our study, we examined the potential of natural AKR1C3 inhibitors in various prostate cancer cell lines and a three-dimensional co-culture spheroid model consisting of cancer cells and cancer-associated fibroblasts (CAFs) mimicking enzalutamide resistant prostate cancer. One of our compounds, named MF-15, expressed strong antineoplastic effects especially in cell culture models with significant enzalutamide resistance. Furthermore, MF-15 exhibited a strong effect on androgen receptor (AR) signaling, including significant inhibition of AR activity, downregulation of androgen-regulated genes, lower prostate specific antigen (PSA) production, and decreased AR and AKR1C3 expression, indicating a bi-functional effect. Even more important, we demonstrated a persisting inhibition of AR activity in the presence of AR-V7 and further showed that MF-15 non-competitively binds within the DNA binding domain of the AR. The data suggest MF-15 as useful drug to overcome enzalutamide resistance.

6.
Chem Biol Interact ; 307: 186-194, 2019 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-31028727

RESUMO

The aldo-keto reductase (AKR) superfamily comprises NAD(P)H-dependent enzymes that catalyze the reduction of a variety of carbonyl compounds. AKRs are classified in families and subfamilies. Humans exhibit three members of the AKR1B subfamily: AKR1B1 (aldose reductase, participates in diabetes complications), AKR1B10 (overexpressed in several cancer types), and the recently described AKR1B15. AKR1B10 and AKR1B15 share 92% sequence identity, as well as the capability of being active towards retinaldehyde. However, AKR1B10 and AKR1B15 exhibit strong differences in substrate specificity and inhibitor selectivity. Remarkably, their substrate-binding sites are the most divergent parts between them. Out of 27 residue substitutions, six are changes to Phe residues in AKR1B15. To investigate the participation of these structural changes, especially the Phe substitutions, in the functional features of each enzyme, we prepared two AKR1B10 mutants. The AKR1B10 m mutant carries a segment of six AKR1B15 residues (299-304, including three Phe residues) in the respective AKR1B10 region. An additional substitution (Val48Phe) was incorporated in the second mutant, AKR1B10mF48. This resulted in structures with smaller and more hydrophobic binding pockets, more similar to that of AKR1B15. In general, the AKR1B10 mutants mirrored well the specific functional features of AKR1B15, i.e., the different preferences towards the retinaldehyde isomers, the much higher activity with steroids and ketones, and the unique behavior with inhibitors. It can be concluded that the Phe residues of loop C (299-304) contouring the substrate-binding site, in addition to Phe at position 48, strongly contribute to a narrower and more hydrophobic site in AKR1B15, which would account for its functional uniqueness. In addition, we have investigated the AKR1B10 and AKR1B15 activity toward steroids. While AKR1B10 only exhibits residual activity, AKR1B15 is an efficient 17-ketosteroid reductase. Finally, the functional role of AKR1B15 in steroid and retinaldehyde metabolism is discussed.


Assuntos
Aldo-Ceto Redutases/metabolismo , Engenharia de Proteínas , Retinoides/metabolismo , Esteroides/metabolismo , Aldo-Ceto Redutases/antagonistas & inibidores , Aldo-Ceto Redutases/genética , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Isomerismo , Cinética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Retinaldeído/química , Retinaldeído/metabolismo , Retinoides/química , Alinhamento de Sequência , Esteroides/química , Especificidade por Substrato
7.
J Steroid Biochem Mol Biol ; 185: 80-89, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30031147

RESUMO

Many enzymes from the short-chain dehydrogenase/reductase superfamily (SDR) have already been well characterized, particularly those that participate in crucial biochemical reactions in the human body (e.g. 11ß-hydroxysteroid dehydrogenase 1, 17ß-hydroxysteroid dehydrogenase 1 or carbonyl reductase 1). Several other SDR enzymes are completely or almost completely uncharacterized, such as DHRS1 (also known as SDR19C1). Based on our in silico and experimental approaches, DHRS1 is described as a likely monotopic protein that interacts with the membrane of the endoplasmic reticulum. The highest expression level of DHRS1 protein was observed in human liver and adrenals. The recombinant form of DHRS1 was purified using the detergent n-dodecyl-ß-D-maltoside, and DHRS1 was proven to be an NADPH-dependent reductase that is able to catalyse the in vitro reductive conversion of some steroids (estrone, androstene-3,17-dione and cortisone), as well as other endogenous substances and xenobiotics. The expression pattern and enzyme activities fit to a role in steroid and/or xenobiotic metabolism; however, more research is needed to fully clarify the exact biological function of DHRS1.


Assuntos
Glândulas Suprarrenais/metabolismo , Retículo Endoplasmático/metabolismo , Fígado/metabolismo , Oxirredutases/genética , Oxirredutases/metabolismo , Redutases-Desidrogenases de Cadeia Curta/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Cortisona/metabolismo , Estrona/metabolismo , Células HeLa , Humanos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Células Sf9
8.
Eur J Med Chem ; 155: 61-76, 2018 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-29859505

RESUMO

The human enzyme 17ß-hydroxysteroid dehydrogenase 14 (17ß-HSD14) oxidizes the hydroxyl group at position 17 of estradiol and 5-androstenediol using NAD+ as cofactor. However, the physiological role of the enzyme remains unclear. We recently described the first class of nonsteroidal inhibitors for this enzyme with compound 1 showing a high 17ß-HSD14 inhibitory activity. Its crystal structure was used as starting point for a structure-based optimization in this study. The goal was to develop a promising chemical probe to further investigate the enzyme. The newly designed compounds revealed mostly very high inhibition of the enzyme and for seven of them the crystal structures of the corresponding inhibitor-enzyme complexes were resolved. The crystal structures disclosed that a small change in the substitution pattern of the compounds resulted in an alternative binding mode for one inhibitor. The profiling of a set of the most potent inhibitors identified 13 (Ki = 9 nM) with a good selectivity profile toward three 17ß-HSDs and the estrogen receptor alpha. This inhibitor displayed no cytotoxicity, good solubility, and auspicious predicted bioavailability. Overall, 13 is a highly interesting 17ß-HSD14 inhibitor, which might be used as chemical probe for further investigation of the target enzyme.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Piridinas/farmacologia , 17-Hidroxiesteroide Desidrogenases/metabolismo , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estrutura Molecular , Piridinas/síntese química , Piridinas/química , Relação Estrutura-Atividade
9.
J Med Chem ; 56(1): 167-81, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23145773

RESUMO

Inhibition of 17ß-HSD2 is an attractive mechanism for the treatment of osteoporosis. We report here the optimization of human 17ß-HSD2 inhibitors in the 2,5-thiophene amide class by varying the size of the linker (n equals 0 and 2) between the amide moiety and the phenyl group. While none of the phenethylamides (n = 2) were active, most of the anilides (n = 0) turned out to moderately or strongly inhibit 17ß-HSD2. The four most active compounds showed an IC50 of around 60 nM and a very good selectivity toward 17ß-HSD1, 17ß-HSD4, 17ß-HSD5, 11ß-HSD1, 11ß-HSD2 and the estrogen receptors α and ß. The investigated compounds inhibited monkey 17ß-HSD2 moderately, and one of them showed good inhibitory activity on mouse 17ß-HSD2. SAR studies allowed a first characterization of the human 17ß-HSD2 active site, which is predicted to be considerably larger than that of 17ß-HSD1.


Assuntos
Amidas/síntese química , Conservadores da Densidade Óssea/síntese química , Estradiol Desidrogenases/antagonistas & inibidores , Osteoporose/tratamento farmacológico , Tiofenos/síntese química , 17-Hidroxiesteroide Desidrogenases , Amidas/química , Amidas/farmacologia , Animais , Conservadores da Densidade Óssea/química , Conservadores da Densidade Óssea/farmacologia , Callithrix , Domínio Catalítico , Linhagem Celular Tumoral , Sistema Livre de Células , Ensaios Enzimáticos , Células HEK293 , Humanos , Camundongos , Microssomos/metabolismo , Modelos Moleculares , Ratos , Relação Estrutura-Atividade , Tiofenos/química , Tiofenos/farmacologia
10.
J Med Chem ; 54(21): 7547-57, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21972996

RESUMO

Estrogen deficiency in postmenopausal women or elderly men is often associated with the skeletal disease osteoporosis. The supplementation of estradiol (E2) in osteoporotic patients is known to prevent bone fracture but cannot be administered because of adverse effect. As 17ß-hydroxysteroid dehydrogenase type 2 (17ß-HSD2) oxidizes E2 to its inactive form estrone (E1) and has been found in osteoblastic cells, it is an attractive target for the treatment of osteoporosis. Twenty-one novel, naphthalene-derived compounds have been synthesized and evaluated for their 17ß-HSD2 inhibition and their selectivity toward 17ß-HSD1 and the estrogen receptors (ERs) α and ß. Compound 19 turned out to be the most potent and selective inhibitor of 17ß-HSD2 in cell-free assays and had a very good cellular activity in MDA-MB-231 cells, expressing naturally 17ß-HSD2. It also showed marked inhibition of the E1-formation by the rat and mouse orthologous enzymes and strong inhibition of monkey 17ß-HSD2. It is thus an appropriate candidate to be further evaluated in a disease-oriented model.


Assuntos
Estradiol Desidrogenases/antagonistas & inibidores , Naftóis/síntese química , Fenóis/síntese química , 17-Hidroxiesteroide Desidrogenases , Animais , Ligação Competitiva , Callithrix , Linhagem Celular Tumoral , Estradiol Desidrogenases/química , Feminino , Humanos , Técnicas In Vitro , Camundongos , Microssomos Hepáticos/enzimologia , Naftóis/química , Naftóis/farmacologia , Fenóis/química , Fenóis/farmacologia , Placenta/enzimologia , Gravidez , Ensaio Radioligante , Ratos , Receptores de Estrogênio/metabolismo , Relação Estrutura-Atividade
11.
OMICS ; 15(5): 325-35, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21332381

RESUMO

Cancer cells have several specific metabolic features, which have been explored for targeted therapies. Agents that promote apoptosis in tumors are currently considered as a powerful tool for cancer therapeutics. The present study aimed to design a fast, reliable and robust system for metabolite measurements in cells lines to observe impact of apoptosis on the metabolome. For that purpose the NBS (newborn screen) mass spectrometry-based metabolomics assay was adapted for cell culture approach. In HEK 293 and in cancer cell lines HepG2, PC3, and MCF7 we searched for metabolic biomarkers of apoptosis differing from that of necrosis. Already nontreated cell lines revealed distinct concentrations of metabolites. Several metabolites indicative for apoptotic processes in cell culture including aspartate, glutamate, methionine, alanine, glycine, propionyl carnitine (C3-carnitine), and malonyl carnitine (C3DC-carnitine) were observed. In some cell lines metabolite changes were visible as early as 4 h after apoptosis induction and preceeding the detection by caspase 3/7 assay. We demonstrated for the first time that the metabolomic signatures might be used in the tests of efficacy of agents causing apoptosis in cell culture. These signatures could be obtained in fast high-throughput screening.


Assuntos
Apoptose , Metaboloma , Neoplasias/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Biomarcadores/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/efeitos da radiação , Células HEK293 , Células Hep G2 , Humanos , Metabolômica , Neoplasias/enzimologia , Estaurosporina/farmacologia
12.
J Steroid Biochem Mol Biol ; 125(1-2): 66-82, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21193039

RESUMO

17ß-Hydroxysteroid dehydrogenases (17ß-HSDs) are oxidoreductases, which play a key role in estrogen and androgen steroid metabolism by catalyzing final steps of the steroid biosynthesis. Up to now, 14 different subtypes have been identified in mammals, which catalyze NAD(P)H or NAD(P)(+) dependent reductions/oxidations at the 17-position of the steroid. Depending on their reductive or oxidative activities, they modulate the intracellular concentration of inactive and active steroids. As the genomic mechanism of steroid action involves binding to a steroid nuclear receptor, 17ß-HSDs act like pre-receptor molecular switches. 17ß-HSDs are thus key enzymes implicated in the different functions of the reproductive tissues in both males and females. The crucial role of estrogens and androgens in the genesis and development of hormone dependent diseases is well recognized. Considering the pivotal role of 17ß-HSDs in steroid hormone modulation and their substrate specificity, these proteins are promising therapeutic targets for diseases like breast cancer, endometriosis, osteoporosis, and prostate cancer. The selective inhibition of the concerned enzymes might provide an effective treatment and a good alternative to the existing endocrine therapies. Herein, we give an overview of functional and structural aspects for the different 17ß-HSDs. We focus on steroidal and non-steroidal inhibitors recently published for each subtype and report on existing animal models for the different 17ß-HSDs and the respective diseases. Article from the Special issue on Targeted Inhibitors.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 17-Hidroxiesteroide Desidrogenases/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , Inibidores Enzimáticos/metabolismo , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , 17-Hidroxiesteroide Desidrogenases/classificação , Sequência de Aminoácidos , Androgênios/química , Androgênios/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Domínio Catalítico , Inibidores Enzimáticos/química , Estrogênios/química , Estrogênios/metabolismo , Feminino , Humanos , Isoenzimas/classificação , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Filogenia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Conformação Proteica , Alinhamento de Sequência
13.
PLoS One ; 5(6): e10969, 2010 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-20544026

RESUMO

Steroid-related cancers can be treated by inhibitors of steroid metabolism. In searching for new inhibitors of human 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD 1) for the treatment of breast cancer or endometriosis, novel substances based on 15-substituted estrone were validated. We checked the specificity for different 17beta-HSD types and species. Compounds were tested for specificity in vitro not only towards recombinant human 17beta-HSD types 1, 2, 4, 5 and 7 but also against 17beta-HSD 1 of several other species including marmoset, pig, mouse, and rat. The latter are used in the processes of pharmacophore screening. We present the quantification of inhibitor preferences between human and animal models. Profound differences in the susceptibility to inhibition of steroid conversion among all 17beta-HSDs analyzed were observed. Especially, the rodent 17beta-HSDs 1 were significantly less sensitive to inhibition compared to the human ortholog, while the most similar inhibition pattern to the human 17beta-HSD 1 was obtained with the marmoset enzyme. Molecular docking experiments predicted estrone as the most potent inhibitor. The best performing compound in enzymatic assays was also highly ranked by docking scoring for the human enzyme. However, species-specific prediction of inhibitor performance by molecular docking was not possible. We show that experiments with good candidate compounds would out-select them in the rodent model during preclinical optimization steps. Potentially active human-relevant drugs, therefore, would no longer be further developed. Activity and efficacy screens in heterologous species systems must be evaluated with caution.


Assuntos
Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Animais , Avaliação Pré-Clínica de Medicamentos , Estradiol Desidrogenases/metabolismo , Humanos , Especificidade da Espécie , Especificidade por Substrato
14.
Bioorg Med Chem Lett ; 19(23): 6740-4, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19836949

RESUMO

In search for specific drugs against steroid-dependent cancers we have developed a novel set of potent inhibitors of steroidogenic human 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD 1). The X-ray structure of 17beta-HSD 1 in complex with estradiol served as basis for the design of the inhibitors. 2-Substituted estrone and D-homo-estrone derivatives were synthesized and tested for 17beta-HSD 1 inhibition. The best 17beta-HSD 1 inhibitor, 2-phenethyl-D-homo-estrone, revealed an IC(50) of 15 nM in vitro. The inhibitory potency of compounds is comparable or better to that of previously described inhibitors. An interaction within the cofactor binding site is not necessary to obtain this high binding affinity for substances developed.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Estrona/síntese química , Estrona/farmacologia , Cristalografia por Raios X , Inibidores Enzimáticos/química , Estrona/análogos & derivados , Humanos , Modelos Moleculares , Estrutura Molecular , Estereoisomerismo , Relação Estrutura-Atividade
15.
J Steroid Biochem Mol Biol ; 117(4-5): 93-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19631742

RESUMO

The enzyme 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1) has become an important drug target for breast cancer because it catalyzes the interconversion of estrone to the biologically more potent estradiol which also plays a crucial role in the etiology of breast cancer. Patients with an increased expression of the 17beta-HSD1 gene have a significantly worse outcome than patients without. Inhibitors for 17beta-HSD1 are therefore included in therapy development. Here we have studied binding of 17beta-HSD1 to substrates and a number of inhibitors using NMR spectroscopy. Ligand observed NMR spectra show a strong pH dependence for the phytoestrogens luteolin and apigenin but not for the natural ligands estradiol and estrone. Moreover, NMR competition experiments show that the phytoestrogens do not replace the estrogens despite their similar inhibition levels in the in vitro assay. These results strongly support an additional 17beta-HSD1 binding site for phytoestrogens which is neither the substrate nor the co-factor binding site. Docking experiments suggest the dimer interface as a possible location. An additional binding site for the phytoestrogens may open new opportunities for the design of inhibitors, not only for 17beta-HSD1, but also for other family members of the short chain dehydrogenases.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Fitoestrógenos/metabolismo , 17-Hidroxiesteroide Desidrogenases/química , 17-Hidroxiesteroide Desidrogenases/genética , Sequência de Bases , Sítios de Ligação , Primers do DNA , Concentração de Íons de Hidrogênio , Ligantes , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular
16.
J Steroid Biochem Mol Biol ; 114(1-2): 72-7, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19444936

RESUMO

The metabolism of steroids at position 17 is catalysed by a growing number of 17beta-hydroxysteroid dehydrogenases (17beta-HSDs). Several human diseases like breast or prostate cancer, endometriosis,metabolic syndrome and mental diseases were associated with dysfunctions of 17beta-HSDs, which consequently became drug targets. This review will focus on identities of 17beta-HSDs and recent advances in analyses of their physiological roles in steroid and lipid metabolism. It will also address the potential of metabolomics in drug development.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Isoenzimas/metabolismo , Esteroides/metabolismo , 17-Hidroxiesteroide Desidrogenases/genética , Animais , Desenho de Fármacos , Humanos , Isoenzimas/genética , Metabolismo dos Lipídeos , Metabolômica , Estrutura Molecular , Esteroides/química
17.
Mol Cell Endocrinol ; 301(1-2): 89-96, 2009 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-19100308

RESUMO

Steroid hormones and their metabolising enzymes have been studied extensively for their potential role in prostate cancer, with more recent interest in the androgen/estrogen inactivating enzyme 17beta-hydroxysteroid dehydrogenase type 4 (HSD17B4). Gene expression profiling showed HSD17B4 to be significantly overexpressed in prostate cancer compared to matched-benign epithelium. We therefore hypothesized that altered HSD17B4 expression may contribute to prostate cancer progression via altered hormone balance. In this study, HSD17B4 mRNA and protein expression were assessed by in situ hybridisation (ISH) and immunohistochemistry (IHC), respectively, in tissue arrays of prostate tissue from 172 patients treated by radical prostatectomy. Overexpression of HSD17B4 mRNA and protein was associated with prostate cancer (P<0.0001) and multivariate Cox proportional hazards analysis, adjusted for known prognostic indicators, demonstrated HSD17B4 mRNA and high protein expression were significant independent predictors of poor patient outcome as measured by time until PSA relapse (mRNA: hazards ratio [HR]=1.90, 95% confidence interval [CI]=1.15-3.12; P<0.0001; and protein: HR=2.09, 95% CI=1.31-3.33; P=0.0026). Here we provide strong evidence that both mRNA and protein overexpression of HSD17B4 is not only associated with the presence of prostate cancer, but is also a significant independent predictor of poor patient outcome.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Biomarcadores Tumorais/metabolismo , Hidroliases/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/terapia , 17-Hidroxiesteroide Desidrogenases/genética , Idoso , Regulação Neoplásica da Expressão Gênica , Humanos , Hidroliases/genética , Imuno-Histoquímica , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , Proteína Multifuncional do Peroxissomo-2 , Modelos de Riscos Proporcionais , Prostatectomia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esteroides/metabolismo , Resultado do Tratamento
18.
Mol Cell Endocrinol ; 248(1-2): 218-24, 2006 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-16406285

RESUMO

In search for new inhibitors of human 17beta-hydroxysteroid dehydrogenase type 1 (h17beta-HSD1) a specific group of steroids with interesting properties including novel compounds was investigated. Several estratriene derivatives with fluorine-substitution in position 17 of the steroidal scaffold were synthesised and tested in vitro towards recombinant h17beta-HSD1, 2, 4, 5 and 7. Moderate, mostly unselective inhibitors of h17beta-HSD1 and h17beta-HSD2 and a selective inhibitor of h17beta-HSD5 were identified. The structure-activity relationship with respect to inhibitory strengths and selectivity of these compounds on five h17beta-HSDs is discussed.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Estrogênios/farmacologia , Flúor/química , 17-Hidroxiesteroide Desidrogenases/química , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Estrenos/síntese química , Estrenos/química , Estrogênios/síntese química , Estrogênios/química , Humanos , Conformação Proteica , Relação Estrutura-Atividade
19.
Mol Endocrinol ; 17(9): 1715-25, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12829805

RESUMO

The protein encoded by the HSD17B7 gene was originally described as a prolactin receptor-associated protein and as 17beta-hydroxysteroid dehydrogenase (HSD) type 7. Its ability to synthesize 17beta-estradiol in vitro has been reported previously. However, we demonstrate that HSD17B7 is the ortholog of the yeast 3-ketosteroid reductase Erg27p and converts zymosterone to zymosterol in vitro, using reduced nicotinamide adenine dinucleotide phosphate as cofactor. Expression of human and murine HSD17B7 in an Erg27p-deficient yeast strain complements the 3-ketosteroid reductase deficiency of the cells and restores growth on sterol-deficient medium. A fusion of HSD17B7 with green fluorescent protein is located in the endoplasmic reticulum, the site of postsqualene cholesterogenesis. Further critical evidence for a role of HSD17B7 in cholesterol metabolism is provided by the observation that its murine ortholog is a member of the same highly distinct embryonic synexpression group as hydroxymethyl-glutaryl-coenzyme A reductase, the rate-limiting enzyme of sterol biogenesis, and is specifically expressed in tissues that are involved in the pathogenesis of congenital cholesterol-deficiency disorders. We conclude that HSD17B7 participates in postsqualene cholesterol biosynthesis, thus completing the molecular cloning of all genes of this central metabolic pathway. In its function as the 3-ketosteroid reductase of cholesterol biosynthesis, HSD17B7 is a novel candidate for inborn errors of cholesterol metabolism.


Assuntos
3-Hidroxiesteroide Desidrogenases/metabolismo , Colesterol/biossíntese , 3-Hidroxiesteroide Desidrogenases/genética , Animais , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/metabolismo , Genes Reporter , Humanos , Camundongos/embriologia , Camundongos/metabolismo , Oxirredutases/genética , Filogenia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
20.
J Neuroimmunol ; 130(1-2): 184-93, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12225901

RESUMO

Stiff-man syndrome (SMS) is a rare autoimmune disorder of the central nervous system associated with autoantibodies to glutamate decarboxylase (GAD). We isolated five brain-reactive human monoclonal antibodies, with reactivity distinct from GAD, from peripheral blood of a patient newly diagnosed with SMS. Two antibodies reacted with both Purkinje cells and ependymal cells, and precipitated an 80-kDa protein from rat neuronal primary cultures, which was also recognized by 12% (3/25) of SMS sera and 13% (2/15) of SMS cerebrospinal fluid (CSF) samples. The corresponding antigen was identified as 17 beta-hydroxysteroid dehydrogenase type 4 and may represent a possible novel target of autoimmunity in SMS.


Assuntos
17-Hidroxiesteroide Desidrogenases/imunologia , Autoanticorpos/imunologia , Sistema Nervoso Central/imunologia , Enoil-CoA Hidratase , Complexos Multienzimáticos , Neurônios/imunologia , Rigidez Muscular Espasmódica/enzimologia , Rigidez Muscular Espasmódica/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Autoanticorpos/sangue , Autoanticorpos/isolamento & purificação , Células Cultivadas , Sistema Nervoso Central/fisiopatologia , Estradiol/metabolismo , Feminino , Feto , Humanos , Hidroliases , Camundongos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Neurônios/enzimologia , Proteína Multifuncional do Peroxissomo-2 , Células de Purkinje/enzimologia , Células de Purkinje/imunologia , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/imunologia , Rigidez Muscular Espasmódica/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA